You are on page 1of 10

Review Article

Human circadian rhythms: physiological and therapeutic relevance


of light and melatonin
Debra J Skene and Josephine Arendt

Abstract
Addresses Ocular light plays a key role in human physiology by transmitting time of day
Centre for Chronobiology, School of information. The production of the pineal gland hormone melatonin is under the
Biomedical and Molecular Sciences, control of the light–dark cycle. Its profile of secretion defines biological night and
University of Surrey, it has been called the ‘darkness hormone’. Light mediates a number of non-
Guildford GU2 7XH, UK
visual responses, such as phase shifting the internal circadian clock, increasing
Correspondence alertness, heart rate and pupil constriction. Both exogenous melatonin and light,
Professor Debra J Skene if appropriately timed, can phase shift the human circadian system. These
E-mail: d.skene@surrey.ac.uk ‘chronobiotic’ effects of light and melatonin have been used successfully to alleviate
and correct circadian rhythm disorders, such as those experienced following travel
This article was prepared at the invitation of across time zones, in night shift work and in circadian sleep disorders. The
the Clinical Sciences Reviews Committee of effectiveness of melatonin and light are currently being optimized in terms of time of
the Association for Clinical Biochemistry.
administration, light intensity, duration and wavelength, and melatonin dose and
formulation. The aim of this review is not to replicate information that has been
reported in a number of reviews of the human circadian timing system and the role
of melatonin and light, but rather to extract findings relevant to the field of clinical
biochemistry.
Ann Clin Biochem 2006; 43: 344–353

Introduction moving from a day shift to a night shift); can be a result


of circadian ‘dysentrainment’ (e.g. circadian rhythm
Most physiological (temperature, hormones, electro- sleep disorders, advanced and delayed sleep phase
lytes) and behavioural (mood, alertness, sleep, perfor- insomnia [ASPS and DSPS]); or can relate to dysfunc-
mance) measures exhibit daily rhythms. Circadian tional photic input to the SCN clock (e.g. in some types
rhythms are de¢ned as those rhythms that are endo- of blindness).
genously generated, have a periodicity of approxi-
mately 24 h and whose rhythmicity persists when The human circadian system
environmental conditions (light, temperature, posture,
etc.) are kept constant. An endogenous circadian Human circadian rhythms
timing system allows an organism to anticipate and Examples of human circadian rhythms are core body
predict daily changes in the environment. Circadian temperature, and secretion of cortisol, melatonin and
rhythms are generated by a pacemaker localized in the thyroid-stimulating hormone (TSH). In normal condi-
hypothalamic suprachiasmatic nuclei (SCN); lesions of tions, these rhythms have a characteristic time of peak
these nuclei produce arrhythmicity. In normal condi- and nadir and there is a set time interval (phase rela-
tions, circadian rhythms are synchronized (entrained) tionship) between the di¡erent rhythms (Figure 1). In
to the 24 h day primarily by the light--dark cycle. the absence of a light--dark cycle (such as experienced
Entrainment to the environmental light--dark cycle by totally blind individuals or in sighted individuals
allows the timing of daily activities (e.g. sleeping and kept in very dim light conditions), circadian rhythms
feeding) to be optimized. Circadian rhythm disorders become desynchronized from the 24 h day and ‘free-
can be caused by a mismatch between external and run’at their intrinsic period length (t). Although there
internal time (e.g. following £ight across time zones, is some debate as to the average period length (t) in

344 r 2006 The Association for Clinical Biochemistry


Human circadian rhythms 345

humans, most studies agree that t is highly individual. di¡erences in light perception and previous light his-
Studies in sighted people in dim light conditions and tory between the study groups.
forced desychrony experiments have reported average
t’s of approximately 24.2 h,1--3 whereas studies in
totally blind people with free-running circadian Measurement of circadian rhythms
rhythms have reported slightly longer t’s of 24.5 h.4--7 In order to establish whether a daily rhythm in a phy-
The reasons for this discrepancy in t are not yet known siological parameter is endogenously generated (circa-
but may relate to methodological di¡erences or the dian) or a result of changes in the environment
(exogenous), or both, ‘constant routine’ protocols are
(a) 60
performed (this is currently considered the gold stan-
dard).8 In constant routine studies, all possible known
concentration (pg/mL)
Plasma melatonin

confounding factors are kept ‘constant’ so that partici-


40
pants are required to remain awake; maintain a con-
stant posture (e.g. semi-recumbent); in constant
20 lighting conditions, usually dim light o10 lux (10 lux
is approximately the light given by a candle 1ft away
0 from an object; it is possible to read and see quite well
in dim light once the eyes have adapted); and receive
(b) 37.1 isocaloric meals hourly for at least 25 h. The contribu-
temperature (οC)

tion of the circadian system (endogenous) and the


environment (exogenous) to an observed rhythm can
Core body

36.9
also be quanti¢ed in ‘forced desynchrony’ experi-
36.7
ments.3 In these experiments, participants live on a 20
or 28 h day for at least 12 days. Forced desynchrony
protocols are designed to be able to separate the in£u-
36.5
ence of the sleep--wake cycle from the in£uence of the
(c) 60 circadian pacemaker.
Although these experiments are extremely demand-
Subjective alertness

ing on both participants and researchers, they reliably


100 = very alert)
(0 = not alert,

40
estimate the circadian component of an observed
rhythm. There are also confounding factors even to
20 forced desynchrony experiments (e.g. scheduling of
sleep length), and because of ¢nancial and resource
0 constraints (e.g. temporal isolation facilities are
required), these experiments are not always possible.
(d) 1.8 Fortunately, measurement of the endogenous melato-
Task performance

nin rhythm provides a relatively reliable surrogate


reaction time (ss)

1.6 way of assessing the timing of the internal circadian


clock in less-controlled conditions provided that cer-
1.4 tain precautions are taken (see later). Measurement of

1.2
Figure 1 Diagrammatic representation of the circadian
(e) 2.0
rhythms of plasma melatonin, core body temperature,
subjective alertness, task performance (reaction time, in
concentration (mmol/L)

1.8 seconds) and triacylglycerol from human beings held in


Triacylglycerol

constant routine conditions (i.e. awake, controlled light,


1.6 posture, activity and meals). The peak in the melatonin rhythm
(panel a), shown by the dotted line, and the low point of the
temperature rhythm (panel b) are within 1 h of each other. The
1.4
low point of the alertness and performance rhythms (panels c
and d, respectively) is shortly after the melatonin peak, and the
1.2 peak in triacylglycerol (panel e) is close to the melatonin peak.
1000 1400 1800 2200 0200 0600 1000 1400 Reproduced with permission from: Rajaratnam SM, Arendt J.
Clock time (h)
Health in a 24-h society. Lancet 2001; 358: 999–100572

Ann Clin Biochem 2006; 43: 344–353


346 Skene and Arendt

melatonin in plasma or saliva -- or measurement of its signal is captured and transmitted to the SCN clock, in
major metabolite 6-sulphatoxymelatonin, usually in addition to other brain structures. This melanopsin
urine -- can enable long-term monitoring of the human network, with some contribution from the rods and
circadian system in laboratory and real-life studies (see cones, is thought to mediate the non-visual e¡ects of
later section). light.20 Ocular light is the major time cue (zeitgeber)
responsible for synchronizing the human circadian
timing system. Although early work suggested that
Molecular clockworks
‘non-photic’ time cues such as meals, exercise and the
In recent years, major progress has been made in un-
sleep--wake cycle were important synchronizers of the
derstanding the molecular mechanism underlying cir-
human circadian clock,21 today light is considered the
cadian oscillations within individual SCN cells. Clock
primary time cue.22 Evidence to support this view has
genes and clock-controlled genes are rhythmically ex-
come from detailed studies of totally blind people, who
pressed primarily via an intracellular transcriptional/
exhibit free-running circadian rhythms in spite of liv-
translational negative feedback loop.9,10 In brief, during
ing with strong social cues.5--7,23 The contribution of
the day, transcription of a number of genes (Per1, Per2,
non-photic time cues to circadian entrainment, how-
Per3, Cry1, Cry2 and Reverba) is activated by hetero-
ever, cannot be ruled out entirely. The demonstration
meric complexes of CLOCK and BMAL1. This transcrip-
of relative co-ordination in blind subjects with
tion continues into the night until nuclear levels of PER
free-running circadian rhythms23,24 and non-photic
and CRY proteins become su⁄ciently high to repress
entrainment in a few blind subjects4 suggests that
CLOCK/BMAL1 activation. Declining levels of PER/
non-photic zeitgebers may have a weak e¡ect on the
CRY in the early morning then allows transcription of
circadian system. At present, however, the non-photic
the genes again and the cycle continues. This constitu-
stimulus or stimuli responsible for these e¡ects have
tes the negative feedback limb of the oscillator core.
not been identi¢ed. The ability of appropriately timed
There is an auxiliary loop driven by Reverba (and possi-
exercise25 or exogenously administered melatonin26
bly Reverbb). The protein products, REVERBa and b,
to shift circadian rhythms represents the best evidence
suppress Bmal1 transcription, disappearance of RE-
to date that indeed non-photic stimuli can a¡ect clock
VERB proteins allowing Bmal1 to peak at the end of
timing. Future studies investigating how photic and
the night (positive limb). Rhythms in clock gene
non-photic stimuli interact to determine circadian
expression have also been characterized in numerous
phase are required.
peripheral tissues (e.g. liver, heart, lung), providing
substantial evidence for the presence of peripheral
clocks.11,12 The question of how these peripheral clocks
interact with the central SCN clock (feedforward/feed- Melatonin
back) and how they are synchronized (directly via
Synthesis and metabolism
hormonal and/or neural pathways and/or indirectly
The pineal hormone melatonin is intimately linked
via behaviour [e.g. feeding]13) is presently a major
with the circadian timing system. The rhythm in mela-
research question.
tonin synthesis is controlled by the SCN via a multi-
synaptic pathway (SCN -- paraventricular nuclei [PVN]
Circadian photoreception -- superior cervical ganglia [SCG]) that terminates in
Considerable advances have also been made regarding the release of noradrenaline from postganglionic SCG
the neuroanatomical pathways involved in the trans- ¢bres in the pineal gland.27 The SCN regulates the
mission of light information from the retina to the melatonin rhythm via GABAergic inhibition and
SCN (circadian photoreception). Both animal14,15 and glutaminergic stimulation of the PVN.28,29 The rate-
human16,17 studies have shown that, in addition to the limiting enzyme in the synthesis of melatonin is aryl-
classical rods and cones, a novel photoreceptor system alkylamine N-acetyltransferase (AANAT). It is likely
is involved in mediating so-called ‘non-image forming’ that the human AANAT enzyme is regulated primarily
visual functions. In addition to image formation, light at a post-transcriptional level, whereas in rodents the
entering the eyes is responsible for a number of non- key event appears to be cyclic adenosine monopho-
image-forming responses, such as circadian resetting sphate (cAMP)-dependent phosphorylation of a tran-
and entrainment, suppression of nocturnal melatonin scription factor that binds to the AANAT promoter.
production, elevation of core body temperature and Rapid decline in activity with light treatment at night
heart rate, increased alertness and performance, and appears to depend on proteasomal proteolysis of AA-
pupil constriction. The discovery of melanopsin,18 a no- NAT following dephosphorylation and removal of a
vel photopigment, and its localization in a network of protective 14-3-3 protein.30,31 (Evidence from both
intrinsically photosensitive retinal ganglion cells,19 structural studies and sequence analyses support the
has provided a basis for understanding how the light notion that the primary function of 14-3-3 proteins lies

Ann Clin Biochem 2006; 43: 344–353


Human circadian rhythms 347

in their preferential binding to phosphorylated sub- studies.33,46--48 The e¡ect of melatonin administration
strates. In humans, there are seven 14-3-3 isoforms (b, on gonadal hormones is more controversial. Most
g, e, z, Z, i, s).)30,31 Melatonin production peaks during the recent studies with near-physiological doses of melato-
dark phase in all species studied to date. Melatonin is nin show no short-term e¡ects on gonadotrophins and
metabolized primarily in the liver via a cytochrome gonadal hormones.49,50 However, altered melatonin se-
P450-mediated hydroxylation into 6-hydroxymelato- cretion has been reported in a number of reproductive
nin. It is then conjugated, primarily with sulphate to pathologies (reviewed byArendt51,52).
form 6-sulphatoxymelatonin and, to a lesser extent,
with glucuronic acid.32,33
Endogenous melatonin rhythm
The circadian rhythm of melatonin has characteristics
that can be used to measure the phase of the circadian
Role of endogenous melatonin
system. In normal 24 h entrained conditions, melato-
The role of endogenous melatonin in human physiol-
nin peaks at 02.00--03.00 h, rising approximately 4 h
ogy is not clearly de¢ned. Although the e¡ects of exo-
earlier (22.00--23.00 h) and declining back to baseline
genous melatonin have been extensively studied
levels in the morning (09.00--10.00 h), while sleep
(mainly at pharmacological doses),26,33,34 more exten-
occurs 1--2 h after melatonin onset, ends 1--2 h before
sive studies with pinealectomized patients and devel-
melatonin o¡set and usually lasts 7 h. The timing of
opment of a speci¢c melatonin receptor antagonist for
the rhythm can be measured by estimating the melato-
human studies may help to clarify its role further.
nin onset, peak or o¡set (Figure 2). Although the time
Endogenous melatonin is a hormonal signal that gives
of melatonin onset in dim light conditions has been
time of day (and time of year) information to the body.
used frequently as a marker of circadian phase, it may
Secreted at night in all species studied to date, it
be better to measure both melatonin onset and o¡set as
appears to reinforce darkness-related behaviours such
there is evidence to suggest that these may be shifted
as sleep initiation. An association between the rise in
di¡erentially.53 Ideally, however, the whole melatonin
melatonin production at night and the increase in sleep
pro¢le should be measured. The amplitude of the
propensity has been noted.35 In addition, abnormal
rhythm and the amount of melatonin produced (e.g.
timing of melatonin in totally blind individuals has
area under the curve) is highly variable between indi-
been associated with increased napping.36 However,
viduals (thought to be related to the size of the pineal
whether these e¡ects of endogenous melatonin are
gland54). However, within an individual, melatonin
causal or merely correlational is not known.
production is fairly consistent.

Melatonin receptors Importance of measuring melatonin


Melatonin receptors are high a⁄nity (picomolar KD) The timing of the melatonin rhythm is today consid-
G-protein (mainly Gi) receptors. Three melatonin ered the most reliable marker of the timing of the circa-
receptors have been cloned, namely mel1a (MT1), mel1b dian clock. Control of the lighting environment and, to
(MT2) and mel1c, by Reppert and colleagues.37--39 a lesser extent, posture, is required for accurate melato-
Although melatonin receptors have been found in the nin measurement but, apart from this, of all possible
human SCN and pituitary stalk,40,41 relatively little ‘rhythm markers’, melatonin is least a¡ected by activity,
work has been done in the area, most likely because of sleep, timing of meals, stress, menstrual cycle, etc. In
the lack of availability of human tissue. Further identi- contrast, other circadian rhythms such as core body
¢cation and localization of melatonin receptors may temperature and secretion of cortisol have been shown
provide some clues as to melatonin’s physiological to be signi¢cantly a¡ected by these factors. Measure-
function in humans. In animals, melatonin receptors ment of melatonin and 6-sulphatoxymelatonin has
in the SCN have been linked to suppression (MT1) and been successfully used to monitor circadian de-
phase shifting (not MT1, possibly MT2) SCN activ- synchrony in blind people in their home environ-
ity.42,43 In sheep, MT1 receptors in the pars tuberalis ments5,55 as well as in DSPS,26 in people working night
mediate the photoperiodic regulation of prolactin pro- shifts56,57 and in people after crossing time zones.26
duction.44 Melatonin receptors in the mediobasal The timing of the melatonin rhythm in these studies
hypothalamus have been proposed to mediate has provided important information as to the indivi-
melatonin’s e¡ects on the gonadal axis of seasonal bree- dual’s circadian phase and has been used to optimize
ders.45 Whether endogenous melatonin has a similar the timing of light and melatonin in the treatment of
role in prolactin production and gonadal development circadian rhythm disorders (discussed later on).
in humans has long been speculated but de¢nitive In addition to being a marker of circadian phase,
proof remains lacking. Exogenous melatonin has been measurement of melatonin is also performed to provide
reported to a¡ect prolactin production in some clinical an assessment of pineal gland function. Conditions

Ann Clin Biochem 2006; 43: 344–353


348 Skene and Arendt

80
Duration
70

60

Plasma melatonin (pg/mL)


Acrophase (calculated peak time)
50 *

40

30 Mid-range crossing
* *
20
25% rise/fall
10 * *
Onset/offset
* *
0
1500 1700 1900 2100 2300 100 300 500 700 900 1100 1300 1500 1700

Clock time (h)

‘Biological night’

Figure 2 Normal profile of melatonin in plasma defining ‘biological night’ based on our laboratory normal values (n ¼ 133, healthy
men and women, all ages). Parameters of this profile and that of salivary melatonin and urinary 6-sulphatoxymelatonin that are used
to characterize the timing of the circadian clock are indicated, namely acrophase, duration, mid-range crossing, 25% rise and fall,
onset and offset of secretion. 6-Sulphatoxymelatonin acrophase is approximately 2 h after the plasma melatonin acrophase.
Redrawn from: Arendt J, Skene DJ. Melatonin as a chronobiotic. Sleep Med Rev 2005; 9: 25–39.26

in which pineal gland function might be compromised are available from a number of manufacturers, includ-
include uveitis, various pineal tumours, disruption of ing Stockgrand Ltd (www.stockgrand.co.uk), ALPCO
neural pathways to the pineal and/or the SCN (e.g. cer- (www.alpco.com) and Buhlmann Laboratories
vical cord lesion, diabetic autonomic neuropathy).33,52 (www.buhlmannlabs.ch). Development of enzyme-
Increased melatonin concentrations have been linked immunosorbent assays (ELISAs) in this area
reported in conditions in which metabolism of melatonin has been slow but ELISAs for saliva melatonin and
is compromised (e.g. liver cirrhosis) and in some repro- 6-sulphatoxymelatonin are currently available.
ductive pathologies (e.g. amenorrhoea, hypogonado-
trophic hypogonadism).33,52 Numerous studies have
reported reduced melatonin production in other condi- Factors affecting melatonin (reviewed by Arendt34)
tions, e.g. breast cancer, sleep disorders (inconsistent), Factors known to a¡ect the timing and production of
depression, heart disease (reviewed byArendt51,52) and melatonin are several. The light--dark cycle has the
Alzheimer’s disease.58 Because of the well-reported in- most signi¢cant a¡ect on melatonin timing and pro-
ter-individual variation in melatonin concentrations as duction. Light is able to advance or delay the timing of
well as an age-related decline,33,58 a cautious approach the melatonin rhythm, depending on when it is admi-
to interpreting cross sectional studies should be nistered. Light at night also has an acute suppressive
adopted. Longitudinal, within-subject studies are pre- e¡ect on melatonin synthesis.61,62 Light transmitted
ferable to determine whether a condition a¡ects pineal via the SCN--PVN--SCG pathway rapidly inhibits the
function or not. activity of AANAT, melatonin’s rate-limiting biosynthetic
enzyme, via proteasomal proteolysis (see previous sec-
tion). Thus, it is essential that the light/dark environ-
Analytical measurement ment is controlled or, if control is impossible, at least
The gold standard for measuring melatonin is gas chro- monitored, when measuring a person’s melatonin con-
matography--mass spectrometry (GCMS).59 For routine centrations. Ideally samples should be collected from
measurement, radioimmunoassays (RIAs) have been subjects in constant dim light conditions (o10 lux at
developed and validated using GCMS and these serve eye level). Posture has also been shown to a¡ect the
as the most popular and widely used way of measuring measurement of plasma melatonin,63 thus ideally this
melatonin and its metabolite, 6-sulphatoxymelatonin. should also be controlled during the sampling period
The current assays and protocols have been recently (routinely subjects are asked to remain seated or semi-
reviewed.60 Assay reagents, antibodies and assay kits recumbent). Drugs reported to interfere with melatonin

Ann Clin Biochem 2006; 43: 344–353


Human circadian rhythms 349

measurements usually a¡ect the synthesis and/or Circadian rhythm disorders


metabolism of melatonin. Melatonin is synthesized
from serotonin via a b1-adrenergic mechanism, and to Circadian rhythm disorders are associated with sleep
a lesser extent an a-adrenergic mechanism. Drugs problems. As such these conditions have been classi¢ed
known to interfere with these processes (e.g. serotonin in the International Classi¢cation of Sleep Disorders
and noradrenaline reuptake inhibitors; a- and b1- ago- (ICSD)70 as Circadian Rhythm Sleep Disorders. Circa-
nists and antagonists) have been shown to a¡ect mela- dian rhythm sleep disorders may be the result of an
tonin concentrations.32--34 Melatonin has been shown abrupt shift in time (e.g. as experienced following
to be metabolized by CYP1A2 in both animals and transmeridian travel), or by an abrupt shift in sleep
humans.64,65 Thus, drugs known to a¡ect the activity and work time (as in rotating shift workers). Sleep dis-
of CYP1A2 (ca¡eine, nicotine) and drugs metabolized turbance is caused by a mismatch between the internal
by CYP1A2 (e.g. £uvoxamine, 5-methoxypsoralen) are circadian timing system and the environment. Follow-
likely to interfere with the metabolism of melatonin. ing a period of re-adaptation, which depends on the
Early ¢ndings reporting high concentrations of melato- number of time zones crossed, synchrony is achieved
nin following £uvoxamine66 and 5-methoxypsoralen67 after time zone travel, but rarely in ‘normal’ night shift
administration can now be explained on the basis of work. Thus, most shift workers are living in a state of
a CYP1A2-mediated mechanism. There is also some intermittent, but chronic desynchrony.71,72 In unusual
preliminary evidence that the in£uence of oral contra- circumstances (social isolation on o¡shore oil rigs
ceptives on melatonin concentrations68,69 may occur at working certain schedules and during the polar win-
the level of melatonin metabolism (unpublished data). ter), most people can completely adapt their internal
Further studies investigating this mechanism are clocks to night work.56,73 Circadian rhythm sleep disor-
required. ders may also be a result of circadian misalignment
such as advanced (ASPS) and delayed sleep phase
insomnia (DSPS), in which su¡erers have abnormally
Ideal sampling conditions early or late bedtimes, respectively. Apart from environ-
The correct procedure to collect blood, saliva and urine mental causes such as lighting at inappropriate times,
for measurement of melatonin and 6-sulphatoxymela- causes of ASPS and DSPS may be intrinsic and relate
tonin is detailed in Table 1. Further details can also be to abnormal light sensitivity or aberrant clock timing
found on the Stockgrand Ltd website (www.stock- or both. Recent evidence suggests that polymorphisms
grand.co.uk) and in a recent review.34 in a number of human clock genes (which could

Table 1 Sampling details for measurement of melatonin and 6-sulphatoxymelatonin (aMT6s)


Biological fluid Analyte Sampling procedure Precautions
Blood Melatonin Collect blood into heparinized tubes. Haemolysed plasma and plasma left in plastic
Centrifuge within 15 min. Store plasma at pipettes for more than 2–3 min may give
201C. falsely elevated melatonin levels.

Saliva Melatonin The best method is to ask subjects to spit Do not eat within 30 min of sampling. Rinse
into polypropylene tubes. Do not stimulate mouth with tap water before spitting. Saliva
saliva production. Store at 201C. left in plastic pipettes for more than 2–3 min
may give falsely elevated melatonin levels.
Salivettes with an untreated cotton plug can Use of salivettes may give falsely elevated
be used. Centrifuge for 15 min at 3000 r.p.m. melatonin levels, standards should be run
Store at 201C. through salivettes to correct for this.

Urine aMT6s Ask subjects to collect all urine passed over a Do not wash urine bottles with bleach or
pre-set period into a standard urine bottle. another oxidant. No preservative is required
Measure and record the volume, store circa 5 mL as aMT6s is stable in urine for 1 day at room
at 201C. Urine should be collected at least temperature, 2 days at 41C and for at least
every 3–4 h (longer during sleep period) for 2 years at 201C.
at least 24 h, preferably for 48 h or longer.
For further details, see www.stockgrand.co.uk

Ann Clin Biochem 2006; 43: 344–353


350 Skene and Arendt

feasibly a¡ect t) may be associated with ASPS However, side-e¡ects such as hangover and headache
(per 2)74,75 and DSPS (per 3).76 Another less-common will need careful monitoring.
circadian rhythm disorder is desynchronization from
the 24 h day as experienced most often in totally blind
people.5,6 There are also some reports of this non-24 h Light
sleep/wake disorder being observed in some sighted Light, appropriately timed, can advance or delay the
people77 and in some dim-light, long-winter environ- circadian timing system. Optimization of light’s phase-
ments,78 although it is rarely seen on British Antarctic shifting e¡ects is currently an intensive area of
Survey bases.79 Non-24 h sleep/wake disorder is cyclic, research. It is clear that the extent to which light can
characterized by periods of good sleep (long duration shift the clock is dependent upon the time of light
sleep with no daytime napping) and periods of poor administration86,87 as well as the intensity of light,88
sleep (short duration sleep with daytime naps36). The its duration and wavelength.53,89,90 Broadly speaking,
periods of poor sleep have been shown to be associated increasing the intensity and duration of the light sti-
with abnormal timing of the circadian clock.23,36 mulus increases its e¡ect. Recent data have shown that
short wavelength light (420--480 nm) is most e¡ective
at phase shifting the human circadian clock.53,89,90 In
Consequences addition an individual’s light history (i.e. previous light
The physiological and behavioural consequences of cir- exposure) may be an important modulator of light’s
cadian misalignment are known to anyone who has phase-shifting e¡ect. Direct proof of this is still out-
travelled across time zones or who has done night shift standing but it is predicted on the basis of reports show-
work: poor sleep of short duration with fatigue and ing light history to a¡ect the ability of light to suppress
reduced alertness, vigilance and performance during melatonin.91--93
waking hours. These symptoms may predispose the
individual to accidents, error and risk. The long-term
consequences of repeated/continuous circadian desyn- Exogenous melatonin
chrony such as may be experienced by shift workers are Appropriately timed, exogenous melatonin has been
just beginning to be studied. There is epidemiological shown to advance or, more controversially, delay the
evidence to show increased cardiovascular risk80,81 timing of the circadian clock.94,95 This phase-shifting
and cancer risk82 in night shift workers. Eating meals e¡ect of melatonin has been employed successfully to
at night has been shown to produce increased blood tri- entrain totally blind people with free-running circa-
glycerides and evidence of insulin resistance,83,84 both dian rhythms.96,97 Entrainment by melatonin in our
risk factors for heart disease. Light exposure at night studies has occurred by melatonin’s ability to phase
has been hypothesized to increase cancer risk.85 advance the circadian clock.96,98 In addition to correct-
Although it is always di⁄cult to relate cause and e¡ect ing the underlying desynchronized circadian disorder,
in chronic studies, these issues clearly require more melatonin has also been shown to improve sleep and
study. reduce daytime napping in these subjects. However,
melatonin also improved sleep in those subjects that
did not entrain (but probably to a lesser extent). Melato-
Treatment strategies nin’s phase-shifting e¡ect is presumably via receptors
The acute and possible long-term consequences of cir-
in the SCN, although this is yet to be de¢nitely proven.
cadian rhythm disorders are driving the development
Current research is directed at determining the mini-
of treatment strategies to alleviate or at least minimize
mum e¡ective dose for entrainment. Melatonin at a dai-
some of the detrimental e¡ects of inappropriate circa-
ly dose of 0.5 mg has been shown to e¡ectively entrain
dian timing (such as tiredness, reduced performance).
free-running rhythms in blind people.98,99 At a single
Currently, light and exogenous melatonins are the best
dose of 0.05 mg, phase shifts can also be seen100 and
candidate treatments as both have been shown to
daily administration of this lower dose has recently
phase shift the timing of the human circadian clock.
been tried in a single blind subject.101 Melatonin has
Although other drug strategies have been used (e.g.
also been successful, when appropriately timed, in the
short acting benzodiazepines, moda¢nil, ca¡eine) to
alleviation of DSPS, jet lag and in helping shift workers
alleviate some of the consequences of circadian misa-
to sleep during the day (reviewed by Arendt and
lignment of jetlag and shift work, whether these drugs
Skene26 and Arendt34).
act directly on the circadian clock is not yet clear. How-
ever, by modifying sleep and wakefulness, these drugs
indirectly a¡ect an individual’s light/dark exposure,
which, in turn, would shift the timing of the clock. It is
Future perspectives
likely that the indirect e¡ect of these drugs on circadian Although manipulation and optimization of the light-
timing will form part of future treatment strategies. ing environment remains key to minimizing the

Ann Clin Biochem 2006; 43: 344–353


Human circadian rhythms 351

detrimental e¡ect of circadian misalignment, the role 8 Duffy JF, Dijk DJ. Getting through to circadian oscillators: why use
of non-photic stimuli in combination with lighting constant routines? J Biol Rhythms 2002; 17: 4–13
requires further study. Timed exercise and meals 9 Reppert SM, Weaver DR. Coordination of circadian timing in
may reinforce the e¡ect of light and help maintain mammals. Nature 2002; 418: 935–41
entrainment. Melatonin is the treatment of choice for 10 Hastings MH, Herzog ED. Clock genes, oscillators, and cellular
networks in the suprachiasmatic nuclei. J Biol Rhythms 2004; 19:
non-24 h sleep/wake disorder experienced by the blind,
400–13
where light treatment is impossible. Further studies
11 Glossop NR, Hardin PE. Central and peripheral circadian
determining the minimum e¡ective dose and the ideal oscillator mechanisms in flies and mammals. J Cell Sci 2002;
dosing regimen (daily/every second day) and formula- 115: 3369–77
tion (fast/slow release) are required. How melatonin’s 12 Yamazaki S, Straume M, Tei H, Sakaki Y, Menaker M, Block GD.
e¡ects are modulated by an individual’s t and pharma- Effects of aging on central and peripheral mammalian clocks.
cokinetics is also not yet clear. Future lighting for Proc Natl Acad Sci USA 2002; 99: 10801–6
industry, institutions and the home needs to incorpo- 13 Schibler U, Ripperger J, Brown SA. Peripheral circadian
rate the recent research developments into practical oscillators in mammals: time and food. J Biol Rhythms 2003;
solutions. The e⁄cacy of lighting optimized on the basis 18: 250–60
of the current research ¢ndings (e.g. optimal spectral 14 Lucas RJ, Freedman MS, Munoz M, Garcia-Fernandez JM,
composition) now needs to be tested in the real-life Foster RG. Regulation of the mammalian pineal by non-rod, non-
situation. In addition, the long-term risk of these cone, ocular photoreceptors. Science 1999; 284: 505–7
developments (e.g. blue light, daily melatonin) needs 15 Lucas RJ, Douglas RH, Foster RG. Characterization of an ocular
careful monitoring. photopigment capable of driving pupillary constriction in mice.
Nat Neurosci 2001; 4: 621–6
16 Brainard GC, Hanifin JP, Greeson JM, et al. Action spectrum for
Acknowledgements melatonin regulation in humans: evidence for a novel circadian
The review was written during the tenure of an EU photoreceptor. J Neurosci 2001; 21: 6405–12
Marie Curie RTN grant (MCRTN-CT-2004-512362) to 17 Thapan K, Arendt J, Skene DJ. An action spectrum for melatonin
suppression: evidence for a novel non-rod, non-cone photo-
DJS, Health and Safety Executive/Energy Institute and
receptor system in humans. J Physiol 2001; 535: 261–7
AFI (Antarctic Funding Initiative CGS-6-15) grants to JA.
18 Provencio I, Jiang G, De Grip WJ, Hayes WP, Rollag MD.
Melanopsin: an opsin in melanophores, brain, and eye. Proc Natl
Acad Sci USA 1998; 95: 340–5
Competing interests: DJS and JA are directors of Stock- 19 Berson DM, Dunn FA, Takao M. Phototransduction by retinal
grand Ltd. ganglion cells that set the circadian clock. Science 2002; 295:
1070–3
20 Hattar S, Lucas RJ, Mrosovsky N, et al. Melanopsin and
References rod–cone photoreceptive systems account for all major accessory
1 Middleton B, Arendt J, Stone BM. Human circadian rhythms in visual functions in mice. Nature 2003; 424: 76–81
constant dim light (8 lux) with knowledge of clock time. J Sleep 21 Aschoff J, Fatranska M, Giedke H, Doerr P, Stamm D, Wisser H.
Res 1996; 5: 69–76 Human circadian rhythms in continuous darkness: entrainment
2 Hiddinga AE, Beersma DG, Van den Hoofdakker RH. by social cues. Science 1971; 171: 213–5
Endogenous and exogenous components in the circadian 22 Czeisler CA. The effect of light on the human circadian pace-
variation of core body temperature in humans. J Sleep Res maker. Ciba Found Symp 1995; 183: 254–90; discussion 90–302.
1997; 6: 156–63 23 Arendt J, Aldhous M, Wright J. Synchronisation of a disturbed
3 Wright Jr KP, Hughes RJ, Kronauer RE, Dijk DJ, Czeisler CA. sleep–wake cycle in a blind man by melatonin treatment. Lancet
Intrinsic near-24-h pacemaker period determines limits of 1988; 1: 772–3
circadian entrainment to a weak synchronizer in humans. Proc 24 Emens JS, Lewy AJ, Lefler BJ, Sack RL. Relative coordination
Natl Acad Sci USA 2001; 98: 14027–32 to unknown ‘weak zeitgebers’ in free-running blind individuals.
4 Klerman EB, Rimmer DW, Dijk DJ, Kronauer RE, Rizzo III JF, J Biol Rhythms 2005; 20: 159–67
Czeisler CA. Nonphotic entrainment of the human circadian 25 Buxton OM, Lee CW, L’Hermite-Baleriaux M, Turek FW, Van
pacemaker. Am J Physiol 1998; 274: R991–6 Cauter E. Exercise elicits phase shifts and acute alterations of
5 Lockley SW, Skene DJ, Arendt J, Tabandeh H, Bird AC, Defrance melatonin that vary with circadian phase. Am J Physiol Regul
R. Relationship between melatonin rhythms and visual loss in the Integr Comp Physiol 2003; 284: R714–24
blind. J Clin Endocrinol Metab 1997; 82: 3763–70 26 Arendt J, Skene DJ. Melatonin as a chronobiotic. Sleep Med Rev
6 Sack RL, Lewy AJ, Blood ML, Keith LD, Nakagawa H. Circadian 2005; 9: 25–39
rhythm abnormalities in totally blind people: incidence and clinical 27 Klein DC, Moore RY. Pineal N-acetyltransferase and hydro-
significance. J Clin Endocrinol Metab 1992; 75: 127–34 xyindole-O-methyltransferase: control by the retinohypothalamic
7 Skene DJ, Lockley SW, Hack LM, Arendt J. Free-running tract and the suprachiasmatic nucleus. Brain Res 1979; 174: 245–62
circadian rhythms in the blind and their correction by melatonin 28 Perreau-Lenz S, Kalsbeek A, Garidou ML, et al. Suprachiasmatic
treatment. In: Honma K, Honma S, eds. Biological Rhythms. control of melatonin synthesis in rats: inhibitory and stimulatory
Sapporo: Hokkaido University Press, 2005; 133–41 mechanisms. Eur J Neurosci 2003; 17: 221–8

Ann Clin Biochem 2006; 43: 344–353


352 Skene and Arendt

29 Perreau-Lenz S, Kalsbeek A, Pevet P, Buijs RM. Glutamatergic 47 Ninomiya T, Iwatani N, Tomoda A, Miike T. Effects of exogenous
clock output stimulates melatonin synthesis at night. Eur J melatonin on pituitary hormones in humans. Clin Physiol 2001;
Neurosci 2004; 19: 318–24 21: 292–9
30 Klein DC, Ganguly S, Coon SL, et al. 14-3-3 proteins in pineal 48 Perras B, Ozcan S, Fehm HL, Born J. Melatonin does not inhibit
photoneuroendocrine transduction: how many roles? J Neuroen- hypothalamic–pituitary–adrenal activity in waking young men.
docrinol 2003; 15: 370–7 J Neuroendocrinol 2005; 17: 811–6
31 Simonneaux V, Ribelayga C. Generation of the melatonin 49 Luboshitzky R, Levi M, Shen-Orr Z, Blumenfeld Z, Herer P, Lavie
endocrine message in mammals: a review of the complex P. Long-term melatonin administration does not alter pituitary–
regulation of melatonin synthesis by norepinephrine, peptides, gonadal hormone secretion in normal men. Hum Reprod 2000;
and other pineal transmitters. Pharmacol Rev 2003; 55: 325–95 15: 60–5
32 Arendt J, Bojkowski C, Franey C, Wright J, Marks V. 50 Rajaratnam SM, Dijk DJ, Middleton B, Stone BM, Arendt J.
Immunoassay of 6-hydroxymelatonin sulfate in human plasma Melatonin phase-shifts human circadian rhythms with no
and urine: abolition of the urinary 24-h rhythm with atenolol.
evidence of changes in the duration of endogenous melatonin
J Clin Endocrinol Metab 1985; 60: 1166–73
secretion or the 24-h production of reproductive hormones. J Clin
33 Arendt J. Melatonin and the Mammalian Pineal Gland. London:
Chapman and Hall, 1995 Endocrinol Metab 2003; 88: 4303–9
34 Arendt J. Melatonin: characteristics, concerns, and prospects. 51 Arendt J. The Pineal Gland and Pineal Tumours. In: Grossman
J Biol Rhythms 2005; 20: 291–303 A, section ed. Endotext.org, Online Endocrinology Textbook
35 Dijk DJ, Cajochen C. Melatonin and the circadian regulation of 2005; http://www.endotext.org/neuroendo/neuroendo15/neuro
sleep initiation, consolidation, structure, and the sleep EEG. endoframe15.htm
J Biol Rhythms 1997; 12: 627–35 52 Arendt J. The Pineal Gland: Basic Physiology and Clinical
36 Lockley SW, Skene DJ, Butler LJ, Arendt J. Sleep and activity Implications. In: DeGroot LJ, Jameson JL, eds. Endocrinology.
rhythms are related to circadian phase in the blind. Sleep 1999; Philadelphia: WB Saunders & Co, 2005
22: 616–23 53 Warman VL, Dijk DJ, Warman GR, Arendt J, Skene DJ. Phase
37 Ebisawa T, Karne S, Lerner MR, Reppert SM. Expression advancing human circadian rhythms with short wavelength light.
cloning of a high-affinity melatonin receptor from Xenopus Neurosci Lett 2003; 342: 37–40
dermal melanophores. Proc Natl Acad Sci USA 1994; 91: 6133–7 54 Chemineau P, Daveau A, Bodin L, Zarazaga L, Gomez-Brunet
38 Reppert SM, Godson C, Mahle CD, Weaver DR, Slaugenhaupt A, Malpaux B. Sheep as a mammalian model of genetic
SA, Gusella JF. Molecular characterization of a second mela- variability in melatonin. Reprod Suppl 2002; 59: 181–90
tonin receptor expressed in human retina and brain: the Mel1b 55 Skene DJ, Lockley SW, Thapan K, Arendt J. Effects of light on
melatonin receptor. Proc Natl Acad Sci USA 1995; 92: 8734–8 human circadian rhythms. Reprod Nutr Dev 1999; 39: 295–304
39 Reppert SM. Melatonin receptors: molecular biology of a new 56 Barnes RG, Deacon SJ, Forbes MJ, Arendt J. Adaptation of the
family of G protein-coupled receptors. J Biol Rhythms 1997; 12: 6-sulphatoxymelatonin rhythm in shiftworkers on offshore oil
528–31 installations during a 2-week 12-h night shift. Neurosci Lett 1998;
40 Weaver DR, Stehle JH, Stopa EG, Reppert SM. Melatonin 241: 9–12
receptors in human hypothalamus and pituitary: implications for 57 Gibbs M, Hampton S, Morgan L, Arendt J. Adaptation of the
circadian and reproductive responses to melatonin. J Clin circadian rhythm of 6-sulphatoxymelatonin to a shift schedule of
Endocrinol Metab 1993; 76: 295–301 seven nights followed by seven days in offshore oil installation
41 Weaver DR, Reppert SM. The Mel1a melatonin receptor gene is workers. Neurosci Lett 2002; 325: 91–4
expressed in human suprachiasmatic nuclei. NeuroReport 1996; 58 Skene DJ, Swaab DF. Melatonin rhythmicity: effect of age and
8: 109–12 Alzheimer’s disease. Exp Gerontol 2003; 38: 199–206
42 Liu C, Weaver DR, Jin X, et al. Molecular dissection of two 59 Lewy AJ, Markey SP. Analysis of melatonin in human plasma by
distinct actions of melatonin on the suprachiasmatic circadian gas chromatography negative chemical ionization mass spectro-
clock. Neuron 1997; 19: 91–102 metry. Science 1978; 201: 741–3
43 Dubocovich ML, Hudson RL, Sumaya IC, Masana MI, Manna E. 60 Middleton B. Melatonin and 6-sulphatoxymelatonin. In: Wheeler
Effect of MT1 melatonin receptor deletion on melatonin-mediated MJ, Morley Hutchinson JS, eds. Hormone Assays in Biological
phase shift of circadian rhythms in the C57BL/6 mouse. J Pineal Fluids. Totowa, NJ: Humana Press, 2005
Res 2005; 39: 113–20 61 Lewy AJ, Wehr TA, Goodwin FK, Newsome DA, Markey SP.
44 Lincoln GA, Andersson H, Hazlerigg D. Clock genes and the long- Light suppresses melatonin secretion in humans. Science 1980;
term regulation of prolactin secretion: evidence for a photoperiod/
210: 1267–9
circannual timer in the pars tuberalis. J Neuroendocrinol 2003; 15:
390–7 62 Bojkowski CJ, Aldhous ME, English J, et al. Suppression of
45 Maywood ES, Bittman EL, Hastings MH. Lesions of the nocturnal plasma melatonin and 6-sulphatoxymelatonin by bright
melatonin- and androgen-responsive tissue of the dorsomedial and dim light in man. Horm Metab Res 1987; 19: 437–40
nucleus of the hypothalamus block the gonadal response of male 63 Deacon S, Arendt J. Posture influences melatonin concen-
Syrian hamsters to programmed infusions of melatonin. Biol trations in plasma and saliva in humans. Neurosci Lett 1994;
Reprod 1996; 54: 470–7 167: 191–4
46 Waldhauser F, Lieberman HR, Lynch HJ, et al. A pharmacolo- 64 Skene DJ, Papagiannidou E, Hashemi E, et al. Contribution of
gical dose of melatonin increases PRL levels in males without CYP1A2 in the hepatic metabolism of melatonin: studies with
altering those of GH, LH, FSH, TSH, testosterone or cortisol. isolated microsomal preparations and liver slices. J Pineal Res
Neuroendocrinology 1987; 46: 125–30 2001; 31: 333–42

Ann Clin Biochem 2006; 43: 344–353


Human circadian rhythms 353

65 Facciola G, Hidestrand M, von Bahr C, Tybring G. Cytochrome 84 Ribeiro DC, Hampton SM, Morgan L, Deacon S, Arendt J.
P450 isoforms involved in melatonin metabolism in human liver Altered postprandial hormone and metabolic responses in a
microsomes. Eur J Clin Pharmacol 2001; 56: 881–8 simulated shift work environment. J Endocrinol 1998; 158:
66 Skene DJ, Bojkowski CJ, Arendt J. Comparison of the effects of 305–10
acute fluvoxamine and desipramine administration on melatonin and 85 Davis S, Mirick DK, Stevens RG. Night shift work, light at night,
cortisol production in humans. Br J Clin Pharmacol 1994; 37: 181–6 and risk of breast cancer. J Natl Cancer Inst 2001; 93: 1557–62
67 Mauviard F, Raynaud F, Geoffriau M, Claustrat B, Pevet P. 5- 86 Minors DS, Waterhouse JM, Wirz-Justice A. A human phase-
Methoxypsoralen inhibits 6-hydroxylation of melatonin in the rat. response curve to light. Neurosci Lett 1991; 133: 36–40
Biol Signals 1995; 4: 32–41 87 Khalsa SB, Jewett ME, Cajochen C, Czeisler CA. A phase
68 Webley GE, Leidenberger F. The circadian pattern of melatonin response curve to single bright light pulses in human subjects.
and its positive relationship with progesterone in women. J Clin J Physiol 2003; 549: 945–52
Endocrinol Metab 1986; 63: 323–8 88 Boivin DB, Duffy JF, Kronauer RE, Czeisler CA. Dose–response
69 Wright Jr KP, Myers BL, Plenzler SC, Drake CL, Badia P. Acute relationships for resetting of human circadian clock by light.
effects of bright light and caffeine on nighttime melatonin and Nature 1996; 379: 540–2
temperature levels in women taking and not taking oral 89 Lockley SW, Brainard GC, Czeisler CA. High Sensitivity of the
contraceptives. Brain Res 2000; 873: 310–7 human circadian melatonin rhythm to resetting by short
70 American Academy of Sleep Medicine. International Classification wavelength light. J Clin Endocrinol Metab 2003; 88: 4502–5
of Sleep disorders: Diagnostic and Coding Manual. Westchester, 90 Revell VL, Arendt J, Terman M, Skene DJ. Short-wavelength
Illinois: American Academy of Sleep Medicine, 2005 sensitivity of the human circadian system to phase-advancing
71 Akerstedt T. Shift work and disturbed sleep/wakefulness. Sleep light. J Biol Rhythms 2005; 20: 270–2
Med Rev 1998; 2: 117–28 91 Owen J, Arendt J. Melatonin suppression in human subjects by
72 Rajaratnam SM, Arendt J. Health in a 24-h society. Lancet 2001; bright and dim light in antarctica: time and season-dependent
358: 999–1005 effects. Neurosci Lett 1992; 137: 181–4
73 Ross JK, Arendt J, Horne J, Haston W. Night-shift work in 92 Hebert M, Martin SK, Lee C, Eastman CI. The effects of prior
Antarctica: sleep characteristics and bright light treatment. light history on the suppression of melatonin by light in humans.
Physiol Behav 1995; 57: 1169–74 J Pineal Res 2002; 33: 198–203
74 Toh KL, Jones CR, He Y, et al. An hPer2 phosphorylation site 93 Smith KA, Schoen MW, Czeisler CA. Adaptation of human pineal
mutation in familial advanced sleep phase syndrome. Science melatonin suppression by recent photic history. J Clin Endocrinol
2001; 291: 1040–3 Metab 2004; 89: 3610–4
75 Carpen JD, Archer SN, Skene DJ, Smits M, von Schantz M. A 94 Lewy AJ, Bauer VK, Ahmed S, et al. The human phase response
single-nucleotide polymorphism in the 50 -untranslated region of curve (PRC) to melatonin is about 12 h out of phase with the
the hPER2 gene is associated with diurnal preference. J Sleep PRC to light. Chronobiol Int 1998; 15: 71–83
Res 2005; 14: 293–7 95 Wirz-Justice A, Werth E, Renz C, Muller S, Krauchi K. No
76 Archer SN, Robilliard DL, Skene DJ, et al. A length evidence for a phase delay in human circadian rhythms after a
polymorphism in the circadian clock gene Per3 is linked to single morning melatonin administration. J Pineal Res 2002; 32:
delayed sleep phase syndrome and extreme diurnal preference. 1–5
Sleep 2003; 26: 413–5 96 Lockley SW, Skene DJ, James K, Thapan K, Wright J, Arendt J.
77 Hayakawa T, Uchiyama M, Kamei Y, et al. Clinical analyses of Melatonin administration can entrain the free-running circadian
sighted patients with non-24-h sleep-wake syndrome: a study of system of blind subjects. J Endocrinol 2000; 164: R1–6
57 consecutively diagnosed cases. Sleep 2005; 28: 945–52 97 Sack RL, Brandes RW, Kendall AR, Lewy AJ. Entrainment of
78 Kennaway DJ, Van Dorp CF. Free-running rhythms of melatonin, free-running circadian rhythms by melatonin in blind people. N
cortisol, electrolytes, and sleep in humans in Antarctica. Am J Engl J Med 2000; 343: 1070–7
Physiol 1991; 260: R1137–44 98 Hack LM, Lockley SW, Arendt J, Skene DJ. The effects of low-
79 Broadway J, Arendt J, Folkard S. Bright light phase shifts the dose 0.5-mg melatonin on the free-running circadian rhythms of
human melatonin rhythm during the Antarctic winter. Neurosci blind subjects. J Biol Rhythms 2003; 18: 420–9
Lett 1987; 79: 185–9 99 Lewy AJ, Bauer VK, Hasler BP, Kendall AR, Pires ML, Sack RL.
80 Knutsson A, Akerstedt T, Jonsson BG, Orth-Gomer K. Increased risk Capturing the circadian rhythms of free-running blind people with
of ischaemic heart disease in shift workers. Lancet 1986; 2: 89–92 0.5 mg melatonin. Brain Res 2001; 918: 96–100
81 Kawachi I, Colditz GA, Stampfer MJ, et al. Prospective study of 100 Deacon S, Arendt J. Melatonin-induced temperature suppression
shift work and risk of coronary heart disease in women. and its acute phase-shifting effects correlate in a dose-
Circulation 1995; 92: 3178–82 dependent manner in humans. Brain Res 1995; 688: 77–85
82 Schernhammer ES, Laden F, Speizer FE, et al. Rotating night 101 Lewy AJ, Emens JS, Bernert RA, Lefler BJ. Eventual
shifts and risk of breast cancer in women participating in the entrainment of the human circadian pacemaker by melatonin is
nurses’ health study. J Natl Cancer Inst 2001; 93: 1563–8 independent of the circadian phase of treatment initiation: clinical
83 Hampton SM, Morgan LM, Lawrence N, et al. Postprandial implications. J Biol Rhythms 2004; 19: 68–75
hormone and metabolic responses in simulated shift work. J
Endocrinol 1996; 151: 259–67 Accepted for publication 22 June 2006

Ann Clin Biochem 2006; 43: 344–353

You might also like